Introduction: The arachidonic acid (AA) pathway plays a crucial role in allergic inflammatory diseases; however, the functional roles of allergy-associated single nucleotide polymorphisms (SNPs) in this pathway remain incompletely illustrated. Methods: This study belongs to a part of an ongoing Singapore/Malaysia cross-sectional genetics and epidemiological study (SMCSGES). We performed population genotyping on n = 2,880 individuals from the SMCSGES cohort to assess the associations of SNPs in the AA pathway genes with asthma and allergic rhinitis (AR). Spirometry assessments were performed to identify associations between SNPs and lung function among n = 74 pediatric asthmatic patients from the same cohort. Allergy-associated SNPs were functionally characterized using in vitro promoter luciferase assay, along with DNA methylome and transcriptome data of n = 237 peripheral blood mononuclear cell (PBMC) samples collected from a subset of the SMCSGES cohort. Results: Genetic association analysis showed 5 tag-SNPs from 4 AA pathway genes were significantly associated with asthma (rs689466 at COX2, rs35744894 at hematopoietic PGD2 synthase (HPGDS), rs11097414 at HPGDS, rs7167 at CRTH2, and rs5758 at TBXA2R, p < 0.05), whereas 3 tag-SNPs from HPGDS (rs35744894, rs11097414, and rs11097411) and 2 tag-SNPs from PTGDR (rs8019916 and rs41312470) were significantly associated with AR (p < 0.05). The asthma-associated rs689466 regulates COX2 promoter activity and associates with COX2 mRNA expression in PBMC. The allergy-associated rs1344612 was significantly associated with poorer lung function, increased risks of asthma and AR, and increased HPGDS promoter activity. The allergy-associated rs8019916 regulates PTGDR promoter activity and DNA methylation levels of cg23022053 and cg18369034 in PBMC. The asthma-associated rs7167 affects CRTH2 expression by regulating the methylation level of cg19192256 in PBMC. Conclusions: The present study identified multiple allergy-associated SNPs that modulate the transcript expressions of key genes in the AA pathway. The development of a “personalized medicine” approach with consideration of genetic influences on the AA pathway may hopefully result in efficacious strategies to manage and treat allergic diseases.

The arachidonic acid (AA) pathway generates a wide spectrum of pro-inflammatory mediators and is identified as one of the key drivers in the development of allergic inflammatory diseases, including asthma and allergic rhinitis (AR) [1, 3]. AA can be metabolized by the cyclooxygenase (COX) enzyme pathway to produce biologically active prostaglandins (PGs) that are important mediators of inflammatory processes [1]. There are two major forms of COXs: COX1 is constitutively expressed in most cells and produces PGs that serve housekeeping functions [4]; COX2 can be either constitutive or inducible, depending on the tissue type. In acute and chronic inflammatory states, COX2 is induced by cytokines to facilitate the synthesis of PGH2. PGH2 is the precursor for prostanoids such as PGD2 and PGE2, as well as thromboxane A2 (TBXA2). PGD2 is mainly produced in mast cells and eosinophils by the hematopoietic PGD2 synthase (HPGDS), which can cause bronchoconstriction, induce vascular leakage, recruits and activates dendritic cells through either the chemoattractant receptor-homologous molecule expressed on T helper 2 cells (CRTH2) or PGD2 receptor (PTGDR) [2, 5]. TBXA2 is synthesized by the TBXA synthase and exerts its biological function via its receptor TBXA2R. Studies have also shown the activation of TBXA2R by its ligand leads to airway smooth muscle cell contraction, proliferation, and hypertrophy [6, 8]. PGE2 can bind to PGE2 receptor 1 (EP1), EP2, EP3, or EP4 and was shown to have protective effect against asthma-related phenotype: inhalation of PGE2 protected against bronchoconstriction [9], while reduced levels of PGE2 were also observed in aspirin-exacerbated respiratory disease (AERD) patients [10].

Globally, allergic diseases including asthma and AR affect about 10–30% of the population [11, 12]. These allergic conditions result from a complex interplay between genetic and environmental factors. Prior studies have investigated the disease associations of single nucleotide polymorphisms (SNPs) in key genes of the AA pathway, including COX2[13, 14], PTGDR[15, 17], CRTH2[18], and TBXA2R[19]. Nevertheless, most findings are considered inconsistent or lack significant disease association. The functional roles of these SNPs in allergic diseases also remain incompletely illustrated. Here, we performed population genotyping in a cohort of n = 2,880 Singaporean Chinese to assess the associations of SNPs in COX2, HPGDS, CRTH2, PTGDR, and TBXA2R with asthma and AR. Allergy-associated SNPs were functionally characterized using DNA methylome and transcriptome data, along with in vitro assays. In the present study, we propose a functional role of allergy-associated genetic polymorphisms in the pathobiology of asthma and AR by modulating the AA pathway.

Study Design and Participant Recruitment

This present study belongs to a part of an ongoing Singapore and Malaysia cross-sectional genetic and epidemiological study (SMCSGES). The recruitment of this cohort has been previously described [20, 23]. Participants were recruited from the National University of Singapore (NUS) from Aug 2005 to Sep 2009, whereas pediatric asthmatic patients were recruited from KK Hospital in Singapore from January 2004 to December 2007. Participants from the University of Tunku Abdul Rahman (UTAR), Malaysia, were recruited from January 2018 to October 2018. The demographics were summarized in Table S1 (for all online suppl. material, see www.karger.com/doi/10.1159/000530393).

An investigator-administered questionnaire was provided to participants to collect information on demographics and allergy-related medical history designed based on the Allergic Rhinitis Impact on Asthma (ARIA) [24] and International Study of Asthma and Allergies in Childhood (ISAAC) [25] guidelines. The Chinese ethnicity of participants was confirmed through survey questionnaires, as well as a previously performed principal component analysis [26]. Atopy was defined as having a positive skin prick test reaction toward common house dust mite allergens (Dermatophagoides pteronyssinus or Blomia tropicalis). A positive skin prick test reaction was defined as having a wheal of at least 3 mm in diameter 15 min after the skin prick. A positive control (1 mg/mL of histamine) and a negative control (0.9% of saline) were also used to validate the allergen-based skin prick measurements. Asthma was defined as ever having asthma positively diagnosed by a physician. AR was defined as having at least two major symptoms that include nasal congestion, rhinorrhea, nasal itching, and sneezing (based on 2008 guidelines set by the ARIA consortium [24]). Both asthma and AR conditions were further confirmed by having an atopy condition. Nonatopic asthma and nonatopic rhinitis individuals were excluded from the analysis. Spirometry measurements were performed on 75 pediatric asthmatic patients from the KK cohort according to American Thoracic Society (ATS) and European Respiratory Society (ERS) guidelines [27].

Genotyping and Sanger Sequencing

Genomic DNA (gDNA) samples were extracted from mouthwash samples for subsequent genotyping and sequencing assays. The extraction was performed using Axygen® AxyPrep™ Multisource Genomic Miniprep DNA Kit (Axygen, USA), according to the manufacturer’s protocol. Quantification of the gDNA concentration was done in triplicates using NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Singapore). Genotyping was performed on the Illumina BeadXpress platform using the GoldenGate Assay (Illumina, Inc., USA) and on the SNP Research Facility at the Washington University in St Louis for genotyping on the Sequenom platform using the MassARRAY iPLEX assay (Sequenom, Inc., USA). For the sequencing of COX2, HPGDS, PTGDR, and TBXA2R, including the 2 kb regions flanking these genes, the process was done via Sanger sequencing, according to published methodology [28].

In vitro Assays

Cell culture: human embryonic kidney cells (HEK293T) were purchased from the American Type Culture Collection (ATCC, USA). HEK293T cells were grown in RPMI-1640 medium (Sigma-Aldrich Co.) supplemented with 10% fetal bovine serum, 2 g/L sodium bicarbonate, and 2 mmol/L l-glutamine. Human keratinocyte cells (HaCat) were purchased from Cell Lines Service (CLS, Germany). HaCat cells were grown in Dulbecco’s modified Eagle’s medium with 10% fetal bovine serum and 3.7 g/L sodium bicarbonate. All cell lines were cultured in a 37 °C humidified incubator with a 5% CO2 environment. Transient transfection of plasmid constructs into the cell lines of interest was performed using Lipofectamine 2000 (Invitrogen, Singapore), with conditions optimized according to the manufacturer’s protocol.

Promoter luciferase assay: the promoter luciferase assay was performed by cloning the putative promoter regions of COX2 (-1,446 bp to -520 bp), HPGDS (-1,562 bp to +332 bp), and PTGDR (1,562 bp to +332 bp) separately into the plasmid pGL4.10 with a promoter-less luciferase gene (Promega, Singapore). Renilla luciferase control plasmid pGL4.74 was also co-transfected for assay normalization purposes. Both Firefly and Renilla luciferase activities were measured using the Dual-Luciferase® Reporter Assay System (Promega, Singapore). Relative luciferase unit was calculated by dividing Firefly luciferase activity reading by that of Renilla luciferase. For the COX2 promoter luciferase assay, testosterone (1 µg/mL), progesterone (1 µg/mL), or DMSO (as a treatment control) was also added at 4 h after transfection into HEK293T. All experiments were performed in triplicates with cells harvested at 24 or 48 h after transfection.

Minigene assay: based on the prediction data from SNPinfo (28), three TBXA2R SNPs (rs1131882, rs4523, and rs5758) are located in the exon splicing enhancer or silencer region of this gene. Thus, we performed an in vitro minigene assay to assess whether these SNPs can affect the splicing activity of TBXA2R. Based on our sequencing data, we identified 3 common haplotypes (frequency >5%) constituted by the rs1131882-rs4523-rs5758 SNP-set, including Hap1 (T-T-C), Hap2 (C-T-C), and Hap3 (C-C-T, online suppl. Fig. S1a). Constructs for the minigene assay were generated to include these 3 haplotypes separately in a 2,952 bp fragment of the TBXA2R gene spanning exon 2–4 of its isoform ß (online suppl. Fig. S1a). The expected length of the transcribed mRNA of TBXA2R isoform a and ß was 2,352 bp and 1,692 bp, respectively. We cloned the 2,952 bp TBXA2R gene fragment into the mammalian expression vector pcDNA3.1/Zeo(+) which has a zeocin resistance gene as an internal transfection control (Invitrogen, Singapore). Constructs were transfected into HEK293T and harvested 24 h after transfection. Total RNA was extracted from the harvested HEK293T cells using E.Z.N.A.® Total RNA Kit (Omega Bio-tek, USA) and converted to cDNA using ProtoScript® First Strand cDNA Synthesis Kit (New England Biolabs); both were performed according to the manufacturer’s protocol. PCR amplification followed by gel electrophoresis was done for both TBXA2R and zeocin cDNA transcripts. The forward and reverse primer sequences used for the PCR amplification of TBXA2R were “TAATACGACTCACTAT” and “TAG?AAG?GCA?CAG?TCG?AGG,” respectively, while the forward and reverse primer sequences used for the PCR amplification of zeocin were “GGC?CAA?GTT?GAC?CAG?TGC?CGT?T” and “CCA?CGA?AGT?GCA?CGC?AGT?T,” respectively. All experiments were performed in triplicates.

Transcriptome Microarray

Approximately 24 mL of whole blood sample was collected from each individual of the SMCSGES cohort, with peripheral blood mononuclear cells (PBMCs) extracted using Ficoll-Hypaque density-gradient centrifugation. Total RNA was extracted from PBMC using E.Z.N.A.® Total RNA Kit (Omega Bio-tek, USA), according to the manufacturer’s protocol. Total RNA samples were sent for whole-transcriptome expression analysis using the Affymetrix Human Exon 1.0 ST Array. The assay was done in a facility provided by Origen Laboratories, Singapore. Affymetrix Power Tools were used for data extraction and quantile normalization, according to reported methodology [29]. Transcript expression data of array probe with transcript cluster ID 2448382 were retrieved for COX2 mRNA expression analysis.

DNA Methylome and Transcriptome Profiling

gDNA and total RNA extraction: for DNA methylome and transcriptome profiling of PBMC samples collected from the SMCSGES cohort, 10 mL of whole blood was collected with PBMCs extracted using Ficoll-Hypaque density-gradient centrifugation. The extraction of gDNA from PBMC was performed using Axygen® AxyPrep™ Multisource Genomic Miniprep DNA Kit (Axygen, USA), while the extraction of total RNA from PBMC was performed using E.Z.N.A.® Total RNA Kit (Omega Bio-tek, USA); both were performed according to the manufacturer’s protocol.

SNP genotyping: the Infinium OmniZhongHua-8 v1.3 BeadChip platform was used for SNP genotyping, according to the manufacturer’s protocols. Haplotype phasing and imputations of the data were performed using the IMPUTE2 program with a reference panel comprising the 1000 Genomes Phase 1 database [30].

DNA methylation profiling: the gDNA samples first underwent the bisulfite conversion process, and the DNA methylome was assayed using the Infinium MethylationEPIC BeadChip Kit (Illumina, San Diego, CA), according to the manufacturer’s protocols. Annotation of the array data was performed using the latest manifest file provided by Illumina, “Infinium MethylationEPIC v1.0 B4 Manifest File,” which comprises information of 865,918 probes. Raw data were preprocessed and quantile normalized, with methylation levels (beta values, ß) subsequently calculated using the Minfi package in the R program version 3.6.1.

Whole-transcriptome sequencing: for library construction, mRNA was first enriched using oligo (dT) beads, and NEBNext Ultra RNA Library Prep Kit was used according to the manufacturer’s protocols. Sequencing was then performed using Illumina NovaSeq 6000 system. Mapping of raw sequence to human genomic sequence (NCBI GRCh38 Assembly) was done using TopHat version 2.1.1. Fragments per kilobase of transcript per million mapped reads were calculated using Cufflinks version 2.2.1. For both DNA methylation profiling and whole-transcriptome sequencing, all samples were measured and analyzed at the same time to prevent batch variation.

Statistical Analysis

The linkage disequilibrium (LD) pattern and r-square values between SNPs were calculated and drawn using software Haploview® vers4.2 [http://www.broadinstitute.org/haploview]. Fisher’s tests were used to calculate genotype-phenotype associations in the PLINK program version 1.06. Fisher’s p, odds ratios, and Hardy-Weinberg equilibrium (HWE) test were all evaluated using the same PLINK program. False discovery rate (FDR) adjustment was performed for all assessed SNPs within each of the candidate genes, based on the Benjamini and Hochberg method [31]. Association analyses of SNP genotypes with normally distributed continuous variables, including mRNA expression levels in PBMC, relative luciferase units measured from the promoter luciferase assay, lung function parameters, and DNA methylation levels (ß), were performed using the independent sample t test or ANOVA test in the R program version 3.6.1 (R Foundation for Statistical Computing, Vienna, Austria). Correlation analyses between DNA methylation levels (ß) and mRNA expression levels in PBMC (fragments per kilobase of transcript per million) were performed using the Pearson’s correlation test in the same R program. Dot plot and box plot figures included in this report were also drawn in the same R program using R package ggplot2 [32].

AA Pathway Genetic Polymorphisms Are Significantly Associated with AA and AR in the Singapore/Malaysia Population

We first aim to identify common SNPs from AA pathway genes (minor allele frequency >0.05) that were presented in the SMCSGES cohort (online suppl. Table S1). We sequenced COX2, HPGDS, PTGDR, and TBXA2R on 40 individuals from this cohort (age = 19.9 ± 1.69, 45% male, online suppl. Table S1) and retrieved genotype information of CRTH2 SNPs from the HapMap Chinese Han (CHB) population database [33]. A total of 38 common SNPs (minor allele frequency >5%) were identified, which were tagged by 21 tag-SNPs using an LD threshold of r2 > 0.8 (online suppl. Fig. S2). Then, 13 tag-SNPs were successfully genotyped on 2,880 individuals from the SMCSGES cohort. Of these, 5 tag-SNPs from 4 genes were significantly associated with asthma (rs689466 at COX2, rs35744894 at HPGDS, rs11097414 at HPGDS, rs7167 at CRTH2, and rs5758 at TBXA2R, p < 0.05, Table 1), whereas 3 tag-SNPs from HPGDS (rs35744894, rs11097414, and rs11097411) and 2 tag-SNPs from PTGDR (rs8019916 and rs41312470) were significantly associated with AR (p < 0.05, Table 2). These SNPs were significantly associated with allergy after FDR adjustment, except for the association between rs7167 and asthma, which was only marginally significant (FDR-adjusted p = 0.066, Table 1, 2). Further, gender-stratified analysis has showed the asthma-associated tag-SNP rs689466 was significant only among the male subjects (n = 1076, FDR-adjusted p = 0.0378) but not among the female subjects (n = 1087, FDR-adjusted p = 0.804, online suppl. Table S2). This suggests sex hormone regulation may be involved in the association between rs689466 and asthma.

Table 1.

Association of tag-SNPs from AA pathway genes with the allergic asthma phenotype

SNPNearbygeneMinor/major allele (A/B)MAFp valuep value (FDR adjusted)OR (95% CI)PHWE
AA versus ABaAA versus BBaAB versus BBa
Asthma versus atopy non-asthma (n = 719vs. n = 1,444) 
rs4648302 COX2 T/G 0.0705 0.984 0.984 0.81 (0.15–4.26) 0.80 (0.15–4.12) 0.98 (0.76–1.27) 0.08592 
rs5275 COX2 C/T 0.188 0.932 0.932 0.96 (0.56–1.65) 0.93 (0.55–1.58) 0.97 (0.80–1.18) 0.1982 
rs689466 COX2 G/A 0.479 0.0107* 0.0321* 1.34 (1.07–1.68) 1.03 (0.81–1.32) 0.77 (0.62–0.95) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0179* 0.0269* 1.57 (1.01–2.42) 1.79 (1.17–2.73) 1.14 (0.94–1.38) 
rs11097414 HPGDS T/A 0.341 0.00609* 0.0183* 1.31 (0.99–1.74) 1.57 (1.18–2.10) 1.20 (0.99–1.46) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0788 0.0788 1.34 (0.97–1.85) 1.45 (1.05–2.00) 1.08 (0.90–1.31) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.545 0.545 1.67 (0.61–4.60) 1.55 (0.57–4.17) 0.93 (0.72–1.19) 0.9071 
rs7167 CRTH2 T/C 0.257 0.118 0.236 0.82 (0.55–1.23) 0.71 (0.48–1.05) 0.87 (0.71–1.05) 0.5206 
rs8019916 PTGDR C/T 0.115 0.979 1.000 0.98 (0.45–2.13) 0.95 (0.45–2.04) 0.98 (0.78–1.23) 0.5207 
rs33998702 PTGDR T/C 0.154 0.120 0.480 0.46 (0.21–1.00) 0.45 (0.21–0.99) 1.00 (0.81–1.22) 0.2821 
rs8004654 PTGDR C/T 0.319 0.271 0.542 0.83 (0.60–1.16) 0.77 (0.55–1.07) 0.92 (0.77–1.12) 0.4907 
rs41312470 PTGDR A/G 0.116 1.000 1.000 0.92 (0.42–1.99) 0.92 (0.43–1.95) 1.00 (0.80–1.26) 0.466 
Asthma versus non-atopy non-asthma (n = 719 vs.n = 716) 
rs4648302 COX2 T/G 0.0705 0.593 0.810 1.80 (0.16–20.17) 2.02 (0.18–22.36) 1.12 (0.83–1.53) 0.08592 
rs5275 COX2 C/T 0.188 0.810 0.810 0.84 (0.45–1.55) 0.89 (0.49–1.62) 1.06 (0.84–1.33) 0.1982 
rs689466 COX2 G/A 0.479 0.642 0.810 1.09 (0.84–1.41) 0.98 (0.74–1.30) 0.90 (0.70–1.15) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0270* 0.0405* 1.63 (0.96–2.78) 1.91 (1.14–3.21) 1.17 (0.94–1.47) 
rs11097414 HPGDS T/A 0.341 0.0117* 0.0351* 1.51 (1.07–2.12) 1.69 (1.19–2.39) 1.12 (0.90–1.40) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0786 0.0786 1.43 (0.97–2.11) 1.55 (1.06–2.28) 1.09 (0.88–1.35) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.988 0.988 1.14 (0.37–3.49) 1.15 (0.39–3.46) 1.02 (0.76–1.36) 0.9071 
rs7167 CRTH2 T/C 0.257 0.0330* 0.0660 0.75 (0.48–1.17) 0.61 (0.39–0.94) 0.82 (0.66–1.02) 0.5206 
rs8019916 PTGDR C/T 0.115 0.520 0.573 1.12 (0.45–2.78) 0.97 (0.40–2.35) 0.86 (0.67–1.12) 0.5207 
rs33998702 PTGDR T/C 0.154 0.182 0.573 0.45 (0.19–1.06) 0.47 (0.20–1.10) 1.05 (0.83–1.33) 0.2821 
rs8004654 PTGDR C/T 0.319 0.328 0.573 0.76 (0.52–1.1) 0.77 (0.53–1.11) 1.01 (0.81–1.26) 0.4907 
rs41312470 PTGDR A/G 0.116 0.573 0.573 1.11 (0.45–2.76) 0.98 (0.40–2.36) 0.88 (0.68–1.13) 0.466 
rs5758 TBXA2R T/C 0.1359 0.0221* 0.0221* 2.67 (1.03–6.92) 3.15 (1.24–8.01) 1.18 (0.91–1.54) 0.8094 
SNPNearbygeneMinor/major allele (A/B)MAFp valuep value (FDR adjusted)OR (95% CI)PHWE
AA versus ABaAA versus BBaAB versus BBa
Asthma versus atopy non-asthma (n = 719vs. n = 1,444) 
rs4648302 COX2 T/G 0.0705 0.984 0.984 0.81 (0.15–4.26) 0.80 (0.15–4.12) 0.98 (0.76–1.27) 0.08592 
rs5275 COX2 C/T 0.188 0.932 0.932 0.96 (0.56–1.65) 0.93 (0.55–1.58) 0.97 (0.80–1.18) 0.1982 
rs689466 COX2 G/A 0.479 0.0107* 0.0321* 1.34 (1.07–1.68) 1.03 (0.81–1.32) 0.77 (0.62–0.95) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0179* 0.0269* 1.57 (1.01–2.42) 1.79 (1.17–2.73) 1.14 (0.94–1.38) 
rs11097414 HPGDS T/A 0.341 0.00609* 0.0183* 1.31 (0.99–1.74) 1.57 (1.18–2.10) 1.20 (0.99–1.46) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0788 0.0788 1.34 (0.97–1.85) 1.45 (1.05–2.00) 1.08 (0.90–1.31) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.545 0.545 1.67 (0.61–4.60) 1.55 (0.57–4.17) 0.93 (0.72–1.19) 0.9071 
rs7167 CRTH2 T/C 0.257 0.118 0.236 0.82 (0.55–1.23) 0.71 (0.48–1.05) 0.87 (0.71–1.05) 0.5206 
rs8019916 PTGDR C/T 0.115 0.979 1.000 0.98 (0.45–2.13) 0.95 (0.45–2.04) 0.98 (0.78–1.23) 0.5207 
rs33998702 PTGDR T/C 0.154 0.120 0.480 0.46 (0.21–1.00) 0.45 (0.21–0.99) 1.00 (0.81–1.22) 0.2821 
rs8004654 PTGDR C/T 0.319 0.271 0.542 0.83 (0.60–1.16) 0.77 (0.55–1.07) 0.92 (0.77–1.12) 0.4907 
rs41312470 PTGDR A/G 0.116 1.000 1.000 0.92 (0.42–1.99) 0.92 (0.43–1.95) 1.00 (0.80–1.26) 0.466 
Asthma versus non-atopy non-asthma (n = 719 vs.n = 716) 
rs4648302 COX2 T/G 0.0705 0.593 0.810 1.80 (0.16–20.17) 2.02 (0.18–22.36) 1.12 (0.83–1.53) 0.08592 
rs5275 COX2 C/T 0.188 0.810 0.810 0.84 (0.45–1.55) 0.89 (0.49–1.62) 1.06 (0.84–1.33) 0.1982 
rs689466 COX2 G/A 0.479 0.642 0.810 1.09 (0.84–1.41) 0.98 (0.74–1.30) 0.90 (0.70–1.15) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0270* 0.0405* 1.63 (0.96–2.78) 1.91 (1.14–3.21) 1.17 (0.94–1.47) 
rs11097414 HPGDS T/A 0.341 0.0117* 0.0351* 1.51 (1.07–2.12) 1.69 (1.19–2.39) 1.12 (0.90–1.40) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0786 0.0786 1.43 (0.97–2.11) 1.55 (1.06–2.28) 1.09 (0.88–1.35) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.988 0.988 1.14 (0.37–3.49) 1.15 (0.39–3.46) 1.02 (0.76–1.36) 0.9071 
rs7167 CRTH2 T/C 0.257 0.0330* 0.0660 0.75 (0.48–1.17) 0.61 (0.39–0.94) 0.82 (0.66–1.02) 0.5206 
rs8019916 PTGDR C/T 0.115 0.520 0.573 1.12 (0.45–2.78) 0.97 (0.40–2.35) 0.86 (0.67–1.12) 0.5207 
rs33998702 PTGDR T/C 0.154 0.182 0.573 0.45 (0.19–1.06) 0.47 (0.20–1.10) 1.05 (0.83–1.33) 0.2821 
rs8004654 PTGDR C/T 0.319 0.328 0.573 0.76 (0.52–1.1) 0.77 (0.53–1.11) 1.01 (0.81–1.26) 0.4907 
rs41312470 PTGDR A/G 0.116 0.573 0.573 1.11 (0.45–2.76) 0.98 (0.40–2.36) 0.88 (0.68–1.13) 0.466 
rs5758 TBXA2R T/C 0.1359 0.0221* 0.0221* 2.67 (1.03–6.92) 3.15 (1.24–8.01) 1.18 (0.91–1.54) 0.8094 

All data were evaluated based on the genotyping results of n = 2,880 individuals from the SMCSGES cohort. For genetic association tests, p value was calculated using the Fisher’s exact test.

FDR adjustment was performed for all assessed SNPs within each of the candidate genes, based on the Benjamini and Hochberg method [31].

The minor allele is designed as “A,” while the major allele is designated as “B” for the SNP.

CI, confidence interval; COX2, cyclooxygenase-2; CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; FDR, false discovery rate; HPGDS, hematopoietic prostaglandin D synthase; MAF, minor allele frequency; OR, odds ratio; PHWE, Hardy-Weinberg equilibrium p value for the tag-SNP assessed in the entire population, with significant cutoff at p < 0.001; PTGDR, prostaglandin D2 receptor; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study; SNP, single nucleotide polymorphism; TBXA2R, thromboxane A2 receptor.

*Fisher’s p < 0.05 is considered as significant.

aReference genotype for the odds ratio calculation.

Table 2.

Association of tag-SNPs from AA pathway genes with the AR phenotype

SNPNearbygeneMinor/major allele (A/B)MAFp valuep value (FDR adjusted)OR (95% CI)PHWE
AA versus ABaAA versus BBaAB versus BBa
AR versus atopy non-AR (n = 795vs. n = 811) 
rs4648302 COX2 T/G 0.0705 0.851 0.851 1.11 (0.22–5.61) 1.00 (0.20–4.99) 0.91 (0.69–1.20) 0.08592 
rs5275 COX2 C/T 0.188 0.715 0.851 1.28 (0.71–2.31) 1.26 (0.71–2.23) 0.98 (0.79–1.21) 0.1982 
rs689466 COX2 G/A 0.479 0.561 0.851 0.88 (0.69–1.13) 0.96 (0.73–1.27) 1.09 (0.86–1.38) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0113* 0.0113* 1.79 (1.06–3.02) 2.07 (1.24–3.45) 1.16 (0.94–1.43) 
rs11097414 HPGDS T/A 0.341 0.00934* 0.0113* 1.41 (1.02–1.95) 1.65 (1.19–2.28) 1.16 (0.94–1.44) 0.6478 
rs11097411 HPGDS G/A 0.291 0.00208* 0.00624* 1.79 (1.23–2.60) 1.91 (1.32–2.77) 1.07 (0.87–1.32) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.372 0.445 1.87 (0.55–6.36) 2.07 (0.62–6.92) 1.11 (0.85–1.44) 0.9071 
rs7167 CRTH2 T/C 0.257 0.445 0.445 1.31 (0.86–1.97) 1.28 (0.85–1.91) 0.98 (0.79–1.2) 0.5206 
rs8019916 PTGDR C/T 0.115 0.00146* 0.00582* 2.84 (1.11–7.22) 1.89 (0.76–4.72) 0.67 (0.52–0.86) 0.5207 
rs33998702 PTGDR T/C 0.154 0.945 0.945 1.06 (0.52–2.16) 1.09 (0.55–2.19) 1.03 (0.82–1.28) 0.2821 
rs8004654 PTGDR C/T 0.319 0.315 0.420 0.99 (0.70–1.39) 0.85 (0.60–1.20) 0.86 (0.70–1.06) 0.4907 
rs41312470 PTGDR A/G 0.116 0.00291* 0.00582* 2.45 (1.00–6.00) 1.65 (0.69–3.97) 0.67 (0.53–0.86) 0.466 
AR versus non-atopy non-AR (n = 795 vs.n = 527) 
rs4648302 COX2 T/G 0.0705 0.337 0.585 1.61 (0.16–15.81) 2.06 (0.21–19.83) 1.28 (0.91–1.79) 0.08592 
rs5275 COX2 C/T 0.188 0.835 0.835 0.90 (0.48–1.68) 0.97 (0.53–1.76) 1.07 (0.84–1.37) 0.1982 
rs689466 COX2 G/A 0.479 0.390 0.585 0.83 (0.63–1.10) 0.94 (0.69–1.27) 1.12 (0.86–1.46) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0932 0.0932 1.72 (0.96–3.11) 1.86 (1.05–3.31) 1.08 (0.85–1.37) 
rs11097414 HPGDS T/A 0.341 0.0380* 0.0570 1.53 (1.06–2.21) 1.61 (1.11–2.34) 1.05 (0.83–1.33) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0151* 0.0453* 1.78 (1.16–2.72) 1.82 (1.19–2.77) 1.02 (0.81–1.29) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.341 0.682 1.50 (0.39–5.84) 1.82 (0.48–6.91) 1.21 (0.90–1.64) 0.9071 
rs7167 CRTH2 T/C 0.257 0.682 0.682 1.08 (0.70–1.68) 0.97 (0.63–1.50) 0.90 (0.71–1.14) 0.5206 
rs8019916 PTGDR C/T 0.115 0.382 0.509 1.37 (0.55–3.39) 1.13 (0.47–2.71) 0.82 (0.62–1.09) 0.5207 
rs33998702 PTGDR T/C 0.154 0.327 0.509 0.98 (0.44–2.21) 1.19 (0.54–2.63) 1.21 (0.94–1.56) 0.2821 
rs8004654 PTGDR C/T 0.319 0.768 0.768 1.00 (0.67–1.50) 1.09 (0.73–1.62) 1.09 (0.86–1.37) 0.4907 
rs41312470 PTGDR A/G 0.116 0.353 0.509 1.37 (0.56–3.40) 1.12 (0.47–2.70) 0.82 (0.62–1.08) 0.466 
SNPNearbygeneMinor/major allele (A/B)MAFp valuep value (FDR adjusted)OR (95% CI)PHWE
AA versus ABaAA versus BBaAB versus BBa
AR versus atopy non-AR (n = 795vs. n = 811) 
rs4648302 COX2 T/G 0.0705 0.851 0.851 1.11 (0.22–5.61) 1.00 (0.20–4.99) 0.91 (0.69–1.20) 0.08592 
rs5275 COX2 C/T 0.188 0.715 0.851 1.28 (0.71–2.31) 1.26 (0.71–2.23) 0.98 (0.79–1.21) 0.1982 
rs689466 COX2 G/A 0.479 0.561 0.851 0.88 (0.69–1.13) 0.96 (0.73–1.27) 1.09 (0.86–1.38) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0113* 0.0113* 1.79 (1.06–3.02) 2.07 (1.24–3.45) 1.16 (0.94–1.43) 
rs11097414 HPGDS T/A 0.341 0.00934* 0.0113* 1.41 (1.02–1.95) 1.65 (1.19–2.28) 1.16 (0.94–1.44) 0.6478 
rs11097411 HPGDS G/A 0.291 0.00208* 0.00624* 1.79 (1.23–2.60) 1.91 (1.32–2.77) 1.07 (0.87–1.32) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.372 0.445 1.87 (0.55–6.36) 2.07 (0.62–6.92) 1.11 (0.85–1.44) 0.9071 
rs7167 CRTH2 T/C 0.257 0.445 0.445 1.31 (0.86–1.97) 1.28 (0.85–1.91) 0.98 (0.79–1.2) 0.5206 
rs8019916 PTGDR C/T 0.115 0.00146* 0.00582* 2.84 (1.11–7.22) 1.89 (0.76–4.72) 0.67 (0.52–0.86) 0.5207 
rs33998702 PTGDR T/C 0.154 0.945 0.945 1.06 (0.52–2.16) 1.09 (0.55–2.19) 1.03 (0.82–1.28) 0.2821 
rs8004654 PTGDR C/T 0.319 0.315 0.420 0.99 (0.70–1.39) 0.85 (0.60–1.20) 0.86 (0.70–1.06) 0.4907 
rs41312470 PTGDR A/G 0.116 0.00291* 0.00582* 2.45 (1.00–6.00) 1.65 (0.69–3.97) 0.67 (0.53–0.86) 0.466 
AR versus non-atopy non-AR (n = 795 vs.n = 527) 
rs4648302 COX2 T/G 0.0705 0.337 0.585 1.61 (0.16–15.81) 2.06 (0.21–19.83) 1.28 (0.91–1.79) 0.08592 
rs5275 COX2 C/T 0.188 0.835 0.835 0.90 (0.48–1.68) 0.97 (0.53–1.76) 1.07 (0.84–1.37) 0.1982 
rs689466 COX2 G/A 0.479 0.390 0.585 0.83 (0.63–1.10) 0.94 (0.69–1.27) 1.12 (0.86–1.46) 0.1441 
rs35744894 HPGDS A/G 0.206 0.0932 0.0932 1.72 (0.96–3.11) 1.86 (1.05–3.31) 1.08 (0.85–1.37) 
rs11097414 HPGDS T/A 0.341 0.0380* 0.0570 1.53 (1.06–2.21) 1.61 (1.11–2.34) 1.05 (0.83–1.33) 0.6478 
rs11097411 HPGDS G/A 0.291 0.0151* 0.0453* 1.78 (1.16–2.72) 1.82 (1.19–2.77) 1.02 (0.81–1.29) 0.3386 
rs530963 CRTH2 G/T 0.0880 0.341 0.682 1.50 (0.39–5.84) 1.82 (0.48–6.91) 1.21 (0.90–1.64) 0.9071 
rs7167 CRTH2 T/C 0.257 0.682 0.682 1.08 (0.70–1.68) 0.97 (0.63–1.50) 0.90 (0.71–1.14) 0.5206 
rs8019916 PTGDR C/T 0.115 0.382 0.509 1.37 (0.55–3.39) 1.13 (0.47–2.71) 0.82 (0.62–1.09) 0.5207 
rs33998702 PTGDR T/C 0.154 0.327 0.509 0.98 (0.44–2.21) 1.19 (0.54–2.63) 1.21 (0.94–1.56) 0.2821 
rs8004654 PTGDR C/T 0.319 0.768 0.768 1.00 (0.67–1.50) 1.09 (0.73–1.62) 1.09 (0.86–1.37) 0.4907 
rs41312470 PTGDR A/G 0.116 0.353 0.509 1.37 (0.56–3.40) 1.12 (0.47–2.70) 0.82 (0.62–1.08) 0.466 

All data were evaluated based on the genotyping results of n = 2,880 individuals from the SMCSGES cohort.

For genetic association tests, p value was calculated using the Fisher’s exact test.

FDR adjustment was performed for all assessed SNPs within each of the candidate genes, based on the Benjamini and Hochberg method (Benjamini and Hochberg 1995 [31]).

The minor allele is designed as “A,” while the major allele is designated as “B” for the SNP.

CI, confidence interval; COX2, cyclooxygenase-2; CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; FDR, false discovery rate; HPGDS, hematopoietic prostaglandin D synthase; MAF, minor allele frequency; OR, odds ratio; PHWE, Hardy-Weinberg equilibrium p value for the tag-SNP assessed in the entire population, with significant cutoff at p < 0.001; PTGDR, prostaglandin D2 receptor; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study; SNP, single nucleotide polymorphism.

*Fisher’s p < 0.05 is considered as significant.

aReference genotype for the odds ratio calculation.

Asthma-Associated Tag-SNP rs689466 Influences Promoter Activity and mRNA Expression of COX2 in the AA Pathway

Next, we assessed if these allergy-associated SNPs have a functional effect on key genes of the AA pathway. An overdominance effect was observed for rs689466 on asthma risk: individuals carrying either genotype “AA” or “GG” have increased risk of the disease, as compared to the “AG” genotype (FDR-adjusted p = 0.0321, OR for AA vs. AG = 1.34, 95% CI = 1.07–1.68; OR for GG vs. AG = 1.30, 95% CI = 1.05–1.61). Given rs689466 is located at -1,192 bp of the COX2 gene, we generated constructs containing -1,446 bp to -520 bp of COX2 to conduct an in vitro luciferase assay. Allele “G” of rs689466 was associated with a significantly higher COX2 promoter activity as compared to allele “A” in HEK293T (p < 0.0001,Fig. 1a). This significant trend was consistently observed even after the addition of sex hormones, including testosterone and progesterone (Fig. 1a). This suggests the gender-specific association of rs689466 with asthma risk might be influenced by other social and environmental factors (reviewed in [34]). Besides, we also used an expression array to measure COX2 transcript expression data of 60 PBMC samples from the SMCSGES cohort (age = 21.28 ± 3.64, 55% male, online suppl. Table S1). Among the male subjects (n = 33), we observed a significant dosage effect of increasing COX2 transcript expression level from genotype “AA” to “GG” of rs689466 (p = 0.0173, Fig. 1b). Concordantly, data from the eQTLgen consortium [35] also showed a significant increase in COX2 mRNA expression in PBMC from allele “A” to “G” of rs689466 (FDR-adjusted p = 1.235x10-23, z-score = 10.021 for allele “G,” online suppl. Table S3).

Fig. 1.

Functional effect of rs689466 on COX2 promoter activities and COX2 mRNA expression levels. a Relative luciferase units (RLUs) of the COX2 promoter constructs with either the allele “A” or “G” of rs689466 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 or 48 h after constructs were transiently transfected into HEK293T. Progesterone (1 µg/mL), testosterone (1 µg/mL), or DMSO (as a treatment control) was also added 4 h after the transfection. Each bar represents the mean RLU ±1 standard deviation of triplicates. bCOX2 mRNA expression levels in PBMC were measured by a transcriptome profiling microarray (probe ID: 2448382) and then compared across rs689466 genotypes among the male subjects (n = 33) in the SMCSGES cohort. Each box represents the mean, ±1 standard deviation, and the minimum and maximum values of the dataset. An independent t test was conducted to determine the statistical significance (p < 0.05) of all associations. COX2, cyclooxygenase-2; PBMCs, peripheral blood mononuclear cells; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study. *p < 0.05, **p < 0.01, ****p < 0.0001.

Fig. 1.

Functional effect of rs689466 on COX2 promoter activities and COX2 mRNA expression levels. a Relative luciferase units (RLUs) of the COX2 promoter constructs with either the allele “A” or “G” of rs689466 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 or 48 h after constructs were transiently transfected into HEK293T. Progesterone (1 µg/mL), testosterone (1 µg/mL), or DMSO (as a treatment control) was also added 4 h after the transfection. Each bar represents the mean RLU ±1 standard deviation of triplicates. bCOX2 mRNA expression levels in PBMC were measured by a transcriptome profiling microarray (probe ID: 2448382) and then compared across rs689466 genotypes among the male subjects (n = 33) in the SMCSGES cohort. Each box represents the mean, ±1 standard deviation, and the minimum and maximum values of the dataset. An independent t test was conducted to determine the statistical significance (p < 0.05) of all associations. COX2, cyclooxygenase-2; PBMCs, peripheral blood mononuclear cells; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study. *p < 0.05, **p < 0.01, ****p < 0.0001.

Close modal

Allergy-Associated Tag-SNP rs11097414 Affects HPGDS Promoter Activity and Associates with Poorer Clinical Outcomes in Pediatric Asthma

The allergy-associated rs11097414 is in a strong LD with rs1344612 (r2 = 1) that is located 1,201 bp upstream of the HPGDS gene. We generated constructs comprising -1,531 bp to +242 bp of the HPGDS gene to conduct an in vitro luciferase assay. Allele “T” of rs1344612 was significantly associated with a higher HPGDS promoter activity as compared to allele “C” in both HEK293T (p < 0.05, Fig. 2a) and HaCat cells (p < 0.05, Fig. 2b). Concordantly, data from the eQTLGen consortium [35] also showed allele “T” of rs1344612 was associated with a higher HPGDS mRNA expression than allele “C” (FDR-adjusted p = 0.00276, z-score = -4.9009 for allele “C,” online suppl. Table S3). This indicates that allele “T” rs1344612 might result in a higher HPGDS transcript expression and upregulated AA pathway, which then contribute to the development of asthma and AR in the SMCSGES cohort.

Fig. 2.

Functional effects of allergy-associated SNPs on HPGDS promoter activity and their correlations with clinical outcomes in pediatric asthma. Relative luciferase units (RLUs) of HEK293T (a) or HaCat (b) cells transfected with HPGDS promoter constructs containing either the allele “C” or “T” of rs1344612 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 or 48 h post-transfection (hpt). Each bar represents the mean RLU ±1 standard deviation of triplicates. Lung function parameters, including the percentages of predicted values of forced mid-expiratory flow (FEF) (c), forced expiratory volume in the first second (FEV1) (d), and FEV1 to forced viral capacity ratio (FEV1/FVC) (e), were measured on 75 pediatric asthmatic patients from the SMCSGES cohort and compared across the genotypes of rs11097414 (a proxy for rs1344612). For each group of genotypes, the mean ±1 standard deviation were shown. An independent t test was conducted to determine the statistical significance (p < 0.05) of associations in (a), (b), (c), (d), and (e). HPGDS, hematopoietic prostaglandin D synthase. *t-test p < 0.05, **t test p < 0.01.

Fig. 2.

Functional effects of allergy-associated SNPs on HPGDS promoter activity and their correlations with clinical outcomes in pediatric asthma. Relative luciferase units (RLUs) of HEK293T (a) or HaCat (b) cells transfected with HPGDS promoter constructs containing either the allele “C” or “T” of rs1344612 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 or 48 h post-transfection (hpt). Each bar represents the mean RLU ±1 standard deviation of triplicates. Lung function parameters, including the percentages of predicted values of forced mid-expiratory flow (FEF) (c), forced expiratory volume in the first second (FEV1) (d), and FEV1 to forced viral capacity ratio (FEV1/FVC) (e), were measured on 75 pediatric asthmatic patients from the SMCSGES cohort and compared across the genotypes of rs11097414 (a proxy for rs1344612). For each group of genotypes, the mean ±1 standard deviation were shown. An independent t test was conducted to determine the statistical significance (p < 0.05) of associations in (a), (b), (c), (d), and (e). HPGDS, hematopoietic prostaglandin D synthase. *t-test p < 0.05, **t test p < 0.01.

Close modal

Besides, among 75 pediatric asthmatic patients in the SMCSGES cohort (age = 10.83 ± 2.67, 72% male, online suppl. Table S1), allele “T” of rs11097414 was also significantly associated with reduced forced expiratory flow (p = 0.00892, Fig. 2c), reduced forced expiratory volume in the first second (FEV1, p = 0.00641, Fig. 2d), and reduced FEV1 to forced vital capacity ratio (p = 0.0295, Fig. 2e). These findings suggested the allele “T” of rs11097414 that was associated with a higher HPGDS transcript expression level might also be associated with poorer clinical outcomes of pediatric asthma.

AR-Associated Tag-SNP rs8019916 Modulates PTGDR Promoter Activity and the Methylation Levels at cg23022053 and cg18369034

The AR-associated rs8019916 is located at -1,005 bp of PTGDR. We generated constructs including -1,520 bp to +396 bp of PTGDR that contain 4 common haplotypes (Hap1, Hap2, Hap3, and Hap4, frequency >5%) formed by 5 SNPs (rs8019916, rs33998702, rs34236606, rs8004654, and rs803010) to conduct an in vitro luciferase assay (Fig. 3a). Allele “C” of rs8019916 was associated with a higher promoter activity of PTGDR in HEK293T (Fig. 3b) and HaCat cells (Fig. 3c). Further, by analyzing DNA methylome and transcriptome data of 237 PBMC samples from the SMCSGES cohort (age = 21.37 ± 4.24, 40.5% male), allele “C” of rs8019916 is also associated with reduced DNA methylation levels at two CpG sites (cg23022053 and cg18369034) located in PTGDR (p = 1.01x10-139 and 7.97x10-13, respectively, Fig. 3d–f). This suggests rs8019916 might influence both promoter activity and DNA methylation levels in PTGDR of the AA pathway, which results in its association with AR susceptibility in the Singapore/Malaysia population.

Fig. 3.

Functional effect of rs8019916 on PTGDR promoter activity and DNA methylation levels. a Frequencies of 4 common haplotypes (Hap1, Hap2, Hap3, and Hap4) formed by 5 SNPs in PTGDR (rs8019916, rs33998702, rs34236606, rs8004654, and rs803010) based on sequencing data of the SMCSGES cohort. b,c Relative luciferase units (RLUs) of HEK293T and HaCat cells transfected with PTGDR promoter constructs containing either Hap1, Hap2, Hap3, or Hap4 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 h after transfection. An independent t test was conducted to determine the statistical significance (p < 0.05) of associations. Each bar represents the mean RLU ±1 standard deviation of triplicates. d Associations between rs8019916 genotype and the methylation levels of 24 CpG sites in PTGDR and the 2 kb flanking region of the gene (chr14: 52,732,416–52,745,442, GRCh37), using 237 PBMC samples from the SMCSGES cohort. DNA methylation levels were assessed as beta values (ß). The ANOVA test was performed and a Bonferroni threshold of p = 0.05/24 = 0.002083 was used to determine the statistical significance of associations. e The methylation levels (ß) of cg23022053 were compared across rs8019916 genotype using n = 237 PBMC samples from the SMCSGES cohort. f Association between DNA methylation levels of cg18369034 and PTGDR mRNA expression levels in n = 237 PBMC samples from the SMCSGES cohort. e,f Each box represents the mean, mean ±1 standard deviation, and the minimum and maximum values of the dataset. The ANOVA test was performed to determine the statistical significance (p < 0.05) of associations in (e) and (f). PTGDR, prostaglandin D2 receptor; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study.

Fig. 3.

Functional effect of rs8019916 on PTGDR promoter activity and DNA methylation levels. a Frequencies of 4 common haplotypes (Hap1, Hap2, Hap3, and Hap4) formed by 5 SNPs in PTGDR (rs8019916, rs33998702, rs34236606, rs8004654, and rs803010) based on sequencing data of the SMCSGES cohort. b,c Relative luciferase units (RLUs) of HEK293T and HaCat cells transfected with PTGDR promoter constructs containing either Hap1, Hap2, Hap3, or Hap4 were compared. Promoter-less luciferase plasmid pGL4.10 serves as a background control. Luciferase activities were measured 24 h after transfection. An independent t test was conducted to determine the statistical significance (p < 0.05) of associations. Each bar represents the mean RLU ±1 standard deviation of triplicates. d Associations between rs8019916 genotype and the methylation levels of 24 CpG sites in PTGDR and the 2 kb flanking region of the gene (chr14: 52,732,416–52,745,442, GRCh37), using 237 PBMC samples from the SMCSGES cohort. DNA methylation levels were assessed as beta values (ß). The ANOVA test was performed and a Bonferroni threshold of p = 0.05/24 = 0.002083 was used to determine the statistical significance of associations. e The methylation levels (ß) of cg23022053 were compared across rs8019916 genotype using n = 237 PBMC samples from the SMCSGES cohort. f Association between DNA methylation levels of cg18369034 and PTGDR mRNA expression levels in n = 237 PBMC samples from the SMCSGES cohort. e,f Each box represents the mean, mean ±1 standard deviation, and the minimum and maximum values of the dataset. The ANOVA test was performed to determine the statistical significance (p < 0.05) of associations in (e) and (f). PTGDR, prostaglandin D2 receptor; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study.

Close modal

Asthma-Associated Tag-SNP rs7167 Influences CRTH2 mRNA Expression in the AA Pathway

The asthma-associated rs7167 is located at 264 bp downstream of the CRTH2. By analyzing the DNA methylome and transcriptome data from the SMCSGES cohort (n = 237, online suppl. Table S1), genotype “TT” of rs7167 was significantly associated with reduced methylation levels of cg19192256 (p = 4.95x10-5), cg12528056 (p = 5.23x10-11), and cg19821841 (p = 0.00105, Fig. 4a, b). Of these CpG sites, the methylation level of cg19192256 was also inversely correlated with the mRNA expression of CRTH2 (Pearson’s R = -0.211, p = 0.00121, Fig. 4c). Besides, data from the eQTLgen consortium [35] showed a significant increase of CRTH2 mRNA expression in PBMC was associated with allele “T” as compared to allele “C” of rs7167 (FDR-adjusted p = 0.00278, z-score = -4.8986 for allele “G,” online suppl. Table S3). These findings suggested genotype “TT” of rs7167 might reduce the methylation level of cg19192256, which result in a higher mRNA expression of CRTH2 and upregulation of AA pathway in PBMC.

Fig. 4.

Functional effect of rs7167 on DNA methylation and CRTH2 mRNA expression levels. a Associations between rs7616 genotype and the methylation levels of 18 CpG sites in CRTH2 and the 2 kb flanking region of the gene (chr11: 60,616,406–60,625,423, GRCh37), using 237 PBMC samples from the SMCSGES cohort. b The methylation levels (ß) of cg19192256 were compared across rs7167 genotypes using n = 237 PBMC samples from the SMCSGES cohort. For each group of genotypes, the mean and ± 1 standard deviation were shown. a,b The ANOVA test was performed and a Bonferroni threshold of p = 0.05/18 = 0.002778 was used to determine the statistical significance (p < 0.002778) of associations. c Correlation between DNA methylation levels of cg19192256 and CRTH2 mRNA expression levels in n = 237 PBMC samples from the SMCSGES cohort was shown. Pearson’s correlation test was performed to determine a significant correlation (p < 0.05). CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; PBMCs, peripheral blood mononuclear cells; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study.

Fig. 4.

Functional effect of rs7167 on DNA methylation and CRTH2 mRNA expression levels. a Associations between rs7616 genotype and the methylation levels of 18 CpG sites in CRTH2 and the 2 kb flanking region of the gene (chr11: 60,616,406–60,625,423, GRCh37), using 237 PBMC samples from the SMCSGES cohort. b The methylation levels (ß) of cg19192256 were compared across rs7167 genotypes using n = 237 PBMC samples from the SMCSGES cohort. For each group of genotypes, the mean and ± 1 standard deviation were shown. a,b The ANOVA test was performed and a Bonferroni threshold of p = 0.05/18 = 0.002778 was used to determine the statistical significance (p < 0.002778) of associations. c Correlation between DNA methylation levels of cg19192256 and CRTH2 mRNA expression levels in n = 237 PBMC samples from the SMCSGES cohort was shown. Pearson’s correlation test was performed to determine a significant correlation (p < 0.05). CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; PBMCs, peripheral blood mononuclear cells; SMCSGES, Singapore/Malaysia cross-sectional genetics and epidemiological study.

Close modal

Asthma-Associated SNP rs5758 Does Not Influence the Splicing Activity of TBXA2R

Based on the prediction data from SNPinfo [36], the asthma-associated rs5758 is in the exon splicing enhancer or silencer region of TBXA2R. Thus, we performed an in vitro minigene assay that focused on a 2,952 bp fragment spanning exon 2–4 of TBXA2R isoform ß (online suppl. Fig. S1a) to assess whether rs5758 can affect the splicing activity of TBXA2R. However, as shown in online supplementary Figure S1b, we did not observe functional effect of rs5758 on the splicing activity of TBXA2R.

The present study reported association of genetic polymorphisms in key genes of the AA pathways with allergy risk and poorer clinical outcomes in pediatric asthma. By analyzing DNA methylome and transcriptome data of PBMC in the SMCSGES cohort, along with in vitro assays, we characterized the functional roles of these polymorphisms in the pathobiology of asthma and AR. Importantly, all of these variants contribute to the manifestation of allergic diseases via modulating the AA pathway (summarized in Fig. 5). These findings highlighted the complexity and importance of this pathway in the development of allergic diseases.

Fig. 5.

Graphical illustration of functional SNPs that were associated with allergy-related phenotype through modulation of the expression of key genes in the AA pathway. All results described in this article were based on in vitro assay on HEK293T and HaCat cells, as well as ex vivo assay on PBMC samples. All allergy-related outcomes resulting from the activations of downstream receptors were discussed in the Introduction section of this article. COX, cyclooxygenase; CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; HPGDS, hematopoietic prostaglandin D synthase; PBMCs, peripheral blood mononuclear cells; mPGES, microsomal PGE synthase; PGD2, prostaglandin D2; PGD2, prostaglandin E2; PGH2, prostaglandin H2; PLA2, phospholipase A2; PTGDR, PGD2 receptor; TBXA2, thromboxane A2; TBXA2R, TBXA2 receptor.

Fig. 5.

Graphical illustration of functional SNPs that were associated with allergy-related phenotype through modulation of the expression of key genes in the AA pathway. All results described in this article were based on in vitro assay on HEK293T and HaCat cells, as well as ex vivo assay on PBMC samples. All allergy-related outcomes resulting from the activations of downstream receptors were discussed in the Introduction section of this article. COX, cyclooxygenase; CRTH2, chemoattractant receptor-homologous molecule expressed on T helper 2 cells; HPGDS, hematopoietic prostaglandin D synthase; PBMCs, peripheral blood mononuclear cells; mPGES, microsomal PGE synthase; PGD2, prostaglandin D2; PGD2, prostaglandin E2; PGH2, prostaglandin H2; PLA2, phospholipase A2; PTGDR, PGD2 receptor; TBXA2, thromboxane A2; TBXA2R, TBXA2 receptor.

Close modal

Our genetic association study focused on a cross-sectional cohort of Singaporean Chinese individuals. In this cohort, the allergic response is dominated by a single class of house dust mite allergen, while nonatopic asthma and nonatopic rhinitis are uncommon [21]. This promotes the detection of allergy-associated genes and reduces false-positive results [37, 38]. In the present study, allele “A” of rs689466 associates with a higher promoter activity and increased mRNA expression of COX2 in the AA pathway. Genotypes “AA” and “GG” of rs689466 are also associated with increased asthma risk among males in our study population (Table 1). Therefore, both upregulation and downregulation of COX2 transcript expression might contribute to the development of this disease differently in the AA pathway. In agreement with this, accumulating evidence has suggested COX2 upregulation can have either beneficial or deleterious effects on the outcome of respiratory diseases, which is largely dependent on the local concentrations of prostanoids produced by COX2 and specific interaction between these prostanoids and their individual receptors (reviewed in [2]). Studies have shown increased expression levels and immunoreactivity of the COX2 enzyme in asthmatic airways [39, 40], while other studies also indicate the pro-inflammatory PGD2, which was synthesized by COX2, has a central role in allergy development and progression [41, 43]. By contrast, reduced COX2 mRNA expression levels were also observed in nasal polyps from aspirin-sensitive asthma patients [44], which then suppressed the synthesis of PGE2 by microsomal PGE synthase and contributed to the development of AERD [2]. A prior study has also shown pro-inflammatory eicosanoids were inversely correlated with atopic sensitization in AERD patients [45].

HPGDS catalyzes the production of PGD2 and is suggested to play a key role in allergic inflammatory responses such as bronchoconstriction, eosinophil survival, and leukocyte activation [41, 43]. PGD2 also has increased levels in patients with severe asthma [46, 47] and in response to allergen challenge [48]. Increased HPGDS expression was observed in the nasal mucosa of AR patients as compared to the controls, while a variety of infiltrating cells expressing HPGDS was also detected in this tissue [49]. In our work, associations between HPGDS expression and risks of allergic diseases were confirmed at the genetic level: the allele “T” of rs11097414 is associated with increased HPGDS promoter activity in HEK293T and HaCat cells while also associated with increased risks of asthma and AR. Besides, the allele “T” of rs11097414 was also associated with poorer clinical outcomes in our pediatric asthma cohort. In line with this, data from prior studies also showed upregulated HPGDS and PGD2 production in bronchial epithelial cells during respiratory syncytial virus bronchiolitis [50], while this infection was associated with an increased risk of childhood asthma [51]. Therefore, pharmacological inhibition of HPGDS and PGD2 biosynthesis might be beneficial to delay the onset of asthma, as well as improve clinical outcomes of this disease. Further, our present findings on the association of rs1107414 with lung function are considered preliminary. Future studies need to validate this by conducting a more comprehensive evaluation of asthma clinical outcomes including lung function test results, asthma control test score, and frequency of asthma symptoms in an independent cohort.

Both PTGDR and CRTH2 are the transmembrane receptors of PGD2 that were frequently proposed as drug targets for the treatment of allergic disease [52, 53]. A genetic study on a multi-ethnic cohort has identified a functional haplotype that was associated with both increased PTGDR promoter activity and increased risk of asthma [54]. Besides, epigenetic changes, such as DNA methylation and histone modifications, have also been frequently identified to associate with allergy susceptibility and development [55, 56]. In line with these findings, the present study has shown rs8019916 influences both promoter activity and DNA methylation levels in the PTGDR gene, which results in its association with allergy.

An association between rs533116 in CRTH2 and allergic asthma was reported in an ethnically diverse population, with evidence showing this may be mediated by the elevated expression of CRTH2[18]. Data also showed allergen-specific CD4+ T cells expressing CRTH2 are increased in the whole blood of allergic subjects as compared to the controls [57]. Although rs533116 was not present in our study population, we identified an asthma-associated SNP rs7167 which also modulates CRTH2 mRNA expression via regulating the DNA methylation level in this region.

There are certain limitations to our study: First, although we demonstrated significant functional effects of asthma-associated SNPs on the promoter activities of AA pathway genes, transcription factors that contributed to these observed effects were not investigated. Further studies using electrophoretic mobility shift assay or chromatin immunoprecipitation assay might help shed light on this question. Second, in this study, the DNA methylome and transcriptome data were assessed in PBMC samples that contain different cell types. This large variability of PBMC samples may result in confounding problems that limit the identification of true positives [55, 58]. Therefore, future studies that utilize single-cell technology for epigenome-wide association study are needed to validate our present findings. Third, although each of the allergy-associated SNPs was shown to be functionally involved in the regulation of AA pathway, we did not observe significant synergistic effect of these SNPs on the development of respiratory allergies. The sample size was too small to perform a subgroup analysis for individuals carrying multiple disease risk alleles, and we plan to address this issue in the future when we have larger numbers of participants, as a part of an ongoing cross-sectional study. Lastly, associations between these SNPs in the AA pathway genes and allergic diseases will also be validated in the future study, using an independent cohort.

In summary, the present study identified multiple allergy-associated SNPs that modulate the transcript expressions of key genes in the AA pathway. Our data highlight the possible molecular mechanisms for how genetic polymorphism in the AA pathway can affect the risk and severity of respiratory allergic diseases. The development of a “personalized medicine” approach with consideration of genetic influences on the AA pathway may hopefully result in efficacious strategies to manage and treat allergy in the future.

We thank all participants and their family members for their willingness to participate in this study and Dr. Ramani Anantharaman, Dr. Bani Kaur Suri, and Parate Pallavi Nilkanth for their help with sample collection.

Participant recruitment study in Singapore was approved by the Institutional Review Board (IRB) in NUS. The study protocol was reviewed and approved by NUS-IRB, approval numbers: 07–023, 09–256, 10–445, 13–075, B-10-343, and H-18-036. Participant recruitment study in Malaysia was approved by the UTAR Scientific and Ethical Review Committee (SERC). The study protocol was reviewed and approved by UTAR-SERC, approval number U/SERC/03/2016. All participants provided informed and written consent, with additional written informed consent obtained from the participants’ parent, legal guardian, or next of kin for those below 21 years old. All recruitment procedures were carried out in concordance with the Helsinki Declaration.

Fook Tim Chew has received consulting fees from Sime Darby Technology Centre, First Resources Ltd., Genting Plantation, and Olam International outside the submitted work. All funding agencies had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. All other authors have no conflicts of interest to declare.

Fook Tim Chew has received research support from the National University of Singapore, Singapore Ministry of Education Academic Research Fund, Singapore Immunology Network, National Medical Research Council (NMRC) (Singapore), Biomedical Research Council (BMRC) (Singapore), National Research Foundation (NRF) (Singapore), Singapore Food Agency (SFA), and the Agency for Science Technology and Research (A*STAR) (Singapore); Grant No.: N-154-000-038-001, R-154-000-191-112, R-154-000-404-112, R-154-000-553-112, R-154-000-565-112, R-154-000-630-112, R-154-000-A08-592, R-154-000-A27-597, R-154-000-A91-592, R-154-000-A95-592, R-154-000-B99-114, BMRC/01/1/21/18/077, BMRC/04/1/21/19/315, BMRC/APG2013/108, SIgN-06-006, SIgN-08-020, NMRC/1150/2008, OFIRG20nov-0033, NRF-MP-2020-0004, SFS_RND_SUFP_001_04, W22W3D0006, H17/01/a0/008, and APG2013/108. Yang Yie SIO has received research support from the NUS Resilience & Growth Postdoctoral Fellowships with Grant No.: R-141-000-036-281. All funding agencies had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

F.T.C. planned and supervised the study. Y.Y.S., P.S., S.A.M., Y.T.R.F., W.C.C., and Y.-H.S. recruited participants for the study. Y.T.R.F. and W.C.C. collected and analyzed the lung function data. Y.Y.S. and P.S. planned and conducted the experiments and analyzed the data. Y.Y.S. wrote the manuscript. All authors reviewed and approved the manuscript.

All data used and included in this study are available from the corresponding author (Chew Fook Tim). Data are not publicly available due to ethical reasons. Further inquiries can be directed to the corresponding author.

1.
Bahia
MS
,
Katare
YK
,
Silakari
O
,
Vyas
B
,
Silakari
P
.
Inhibitors of microsomal prostaglandin E2 synthase-1 enzyme as emerging anti-inflammatory candidates
.
Med Res Rev
.
2014 Jul
34
4
825
55
.
2.
Rumzhum
NN
,
Ammit
AJ
.
Cyclooxygenase 2: its regulation, role and impact in airway inflammation
.
Clin Exp Allergy
.
2016 Mar
46
3
397
410
.
3.
Wang
B
,
Wu
L
,
Chen
J
,
Dong
L
,
Chen
C
,
Wen
Z
.
Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets
.
Signal Transduct Target Ther
.
2021 Feb 26
6
1
94
.
4.
Dubois
RN
,
Abramson
SB
,
Crofford
L
,
Gupta
RA
,
Simon
LS
,
Putte
LBA
.
Cyclooxygenase in biology and disease
.
FASEB J
.
1998 Sep
12
12
1063
73
.
5.
Claar
D
,
Hartert
TV
,
Peebles
RS
Jr
.
The role of prostaglandins in allergic lung inflammation and asthma
.
Expert Rev Respir Med
.
2015 Feb
9
1
55
72
.
6.
Capra
V
,
Habib
A
,
Accomazzo
MR
,
Ravasi
S
,
Citro
S
,
Levy-Toledano
S
.
Thromboxane prostanoid receptor in human airway smooth muscle cells: a relevant role in proliferation
.
Eur J Pharmacol
.
2003 Aug 8
474
2–3
149
59
.
7.
Suzuki
Y
,
Asano
K
,
Shiraishi
Y
,
Oguma
T
,
Shiomi
T
,
Fukunaga
K
.
Human bronchial smooth muscle cell proliferation via thromboxane A2 receptor
.
Prostaglandins Leukot Essent Fatty Acids
.
2004 Dec
71
6
375
82
.
8.
Rolin
S
,
Masereel
B
,
Dogne
JM
.
Prostanoids as pharmacological targets in COPD and asthma
.
Eur J Pharmacol
.
2006 Mar 8
533
1–3
89
100
.
9.
Pavord
ID
,
Tattersfield
AE
.
Bronchoprotective role for endogenous prostaglandin E2
.
Lancet
.
1995 Feb 18
345
8947
436
8
.
10.
Laidlaw
TM
,
Boyce
JA
.
Pathogenesis of aspirin-exacerbated respiratory disease and reactions
.
Immunol Allergy Clin North Am
.
2013 May
33
2
195
210
.
11.
The global asthma report 2018
Auckland, New Zealand
Global Asthma Network
2018
.
12.
Savoure
M
,
Bousquet
J
,
Jaakkola
JJK
,
Jaakkola
MS
,
Jacquemin
B
,
Nadif
R
.
Worldwide prevalence of rhinitis in adults: a review of definitions and temporal evolution
.
Clin Transl Allergy
.
2022 Mar
12
3
e12130
.
13.
Shi
J
,
Misso
NL
,
Duffy
DL
,
Thompson
PJ
,
Kedda
MA
.
A functional polymorphism in the promoter region of the cyclooxygenase-2 gene is not associated with asthma and atopy in an Australian population
.
Clin Exp Allergy
.
2004 Nov
34
11
1714
8
.
14.
Szczeklik
W
,
Sanak
M
,
Szczeklik
A
.
Functional effects and gender association of COX-2 gene polymorphism G-765C in bronchial asthma
.
J Allergy Clin Immunol
.
2004 Aug
114
2
248
53
.
15.
Tsai
YJ
,
Choudhry
S
,
Kho
J
,
Beckman
K
,
Tsai
HJ
,
Navarro
D
.
The PTGDR gene is not associated with asthma in 3 ethnically diverse populations
.
J Allergy Clin Immunol
.
2006 Dec
118
6
1242
8
.
16.
Zhu
G
,
Vestbo
J
,
Lenney
W
,
Silverman
M
,
Whyte
M
,
Helms
P
.
Association of PTGDR gene polymorphisms with asthma in two Caucasian populations
.
Genes Immun
.
2007 Jul
8
5
398
403
.
17.
Leung
TF
,
Li
CY
,
Kong
AP
,
Chan
IH
,
Ng
MC
,
Chan
MH
.
PTGDR is not a major candidate gene for asthma and atopy in Chinese children
.
Pediatr Allergy Immunol
.
2009 Sep
20
6
556
62
.
18.
Campos Alberto
E
,
Maclean
E
,
Davidson
C
,
Palikhe
NS
,
Storie
J
,
Tse
C
.
The single nucleotide polymorphism CRTh2 rs533116 is associated with allergic asthma and increased expression of CRTh2
.
Allergy
.
2012 Nov
67
11
1357
64
.
19.
Pan
Y
,
Li
S
,
Xie
X
,
Li
M
.
Association between thromboxane A2 receptor polymorphisms and asthma risk: a meta-analysis
.
J Asthma
.
2016 Aug
53
6
576
82
.
20.
Andiappan
AK
,
Wang
DY
,
Anantharaman
R
,
Parate
PN
,
Suri
BK
,
Low
HQ
.
Genome-wide association study for atopy and allergic rhinitis in a Singapore Chinese population
.
PLoS One
.
2011
;
6
(
5
):
e19719
.
21.
Andiappan
AK
,
Puan
KJ
,
Lee
B
,
Nardin
A
,
Poidinger
M
,
Connolly
J
.
Allergic airway diseases in a tropical urban environment are driven by dominant mono-specific sensitization against house dust mites
.
Allergy
.
2014 Apr
69
4
501
9
.
22.
Lim
JJ
,
Lim
YYE
,
Ng
JY
,
Malipeddi
P
,
Ng
YT
,
Teo
WY
.
An update on the prevalence, chronicity, and severity of atopic dermatitis and the associated epidemiological risk factors in the Singapore/Malaysia Chinese young adult population: a detailed description of the Singapore/Malaysia Cross-Sectional Genetics Epidemiology Study (SMCGES) cohort
.
World Allergy Organ J
.
2022 Dec
15
12
100722
.
23.
Wong
QYA
,
Lim
JJ
,
Ng
JY
,
Malipeddi
P
,
Teo
WY
,
Lim
YYE
.
Allergic rhinitis in Chinese young adults from the Singapore/Malaysia cross-sectional genetics epidemiology study (SMCGES) cohort: prevalence, patterns, and epidemiology of allergic rhinitis
.
World Allergy Organ J
.
2022 Oct
15
10
100704
.
24.
Bousquet
J
,
Khaltaev
N
,
Cruz
AA
,
Denburg
J
,
Fokkens
WJ
,
Togias
A
.
Allergic rhinitis and its impact on asthma (ARIA) 2008 update (in collaboration with the world health organization, GA(2)len and AllerGen)
.
Allergy
.
2008 Apr
63
Suppl 86
8
160
.
25.
Asher
MI
,
Keil
U
,
Anderson
HR
,
Beasley
R
,
Crane
J
,
Martinez
F
.
International study of asthma and Allergies in childhood (ISAAC): rationale and methods
.
Eur Respir J
.
1995 Mar
8
3
483
91
.
26.
Andiappan
AK
,
Anantharaman
R
,
Nilkanth
PP
,
Wang
DY
,
Chew
FT
.
Evaluating the transferability of Hapmap SNPs to a Singapore Chinese population
.
BMC Genet
.
2010
;
11
:
36
.
27.
Miller
MR
,
Hankinson
J
,
Brusasco
V
,
Burgos
F
,
Casaburi
R
,
Coates
A
.
Standardisation of spirometry
.
Eur Respir J
.
2005 Aug
26
2
319
38
.
28.
Sanger
F
,
Nicklen
S
,
Coulson
AR
.
DNA sequencing with chain-terminating inhibitors
.
Proc Natl Acad Sci U S A
.
1977 Dec
74
12
5463
7
.
29.
Lockstone
HE
.
Exon array data analysis using Affymetrix power tools and R statistical software
.
Brief Bioinform
.
2011 Nov
12
6
634
44
.
30.
1000 Genomes Project Consortium
Auton
A
,
Brooks
LD
,
Durbin
RM
,
Garrison
EP
,
Kang
HM
.
A global reference for human genetic variation
.
Nature
.
2015 Oct 01
526
7571
68
74
.
31.
Benjamini
Y
,
Hochberg
Y
.
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J Roy Stat Soc Ser B Methodol
.
1995
;
57
(
1
):
289
300
.
32.
Wickham
H
ggplot2 : elegant graphics for data analysis
New York ; London
Springer
2009
.
33.
The International HapMap Consortium
.
The international HapMap project
.
Nature
.
2003 Dec 18
426
6968
789
96
.
34.
Chowdhury
NU
,
Guntur
VP
,
Newcomb
DC
,
Wechsler
ME
.
Sex and gender in asthma
.
Eur Respir Rev
.
2021 Dec 31
30
162
210067
.
35.
Võsa
U
,
Claringbould
A
,
Westra
H-J
,
Bonder
MJ
,
Deelen
P
,
Zeng
B
.
Unraveling the polygenic architecture of complex traits using blood eQTL metaanalysis
.
bioRxiv
.
2018
447367
.
36.
Xu
Z
,
Taylor
JA
.
SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies
.
Nucleic Acids Res
.
2009 Jul
37
Web Server issue
W600
5
.
37.
Sio
YY
,
Matta
SA
,
Ng
YT
,
Chew
FT
.
Epistasis between phenylethanolamine N-methyltransferase and β2-adrenergic receptor influences extracellular epinephrine level and associates with the susceptibility to allergic asthma
.
Clin Exp Allergy
.
2020
;
50
(
3
):
352
63
.
38.
Sio
YY
,
Shi
P
,
Say
YH
,
Chew
FT
.
Functional variants in the chromosome 4q21 locus contribute to allergic rhinitis risk by modulating the expression of N-acylethanolamine acid amidase
.
Clin Exp Allergy
.
2022 Jan
52
1
127
36
.
39.
Sousa
A
,
Pfister
R
,
Christie
PE
,
Lane
SJ
,
Nasser
SM
,
Schmitz-Schumann
M
.
Enhanced expression of cyclo-oxygenase isoenzyme 2 (COX-2) in asthmatic airways and its cellular distribution in aspirin-sensitive asthma
.
Thorax
.
1997 Nov
52
11
940
5
.
40.
Chambers
LS
,
Black
JL
,
Ge
Q
,
Carlin
SM
,
Au
WW
,
Poniris
M
.
PAR-2 activation, PGE2, and COX-2 in human asthmatic and nonasthmatic airway smooth muscle cells
.
Am J Physiol Lung Cell Mol Physiol
.
2003 Sep
285
3
L619
27
.
41.
Pettipher
R
.
The roles of the prostaglandin D(2) receptors DP(1) and CRTH2 in promoting allergic responses
.
Br J Pharmacol
.
2008 Mar
153
Suppl 1
S191
9
.
42.
Marone
G
,
Galdiero
MR
,
Pecoraro
A
,
Pucino
V
,
Criscuolo
G
,
Triassi
M
.
Prostaglandin D2 receptor antagonists in allergic disorders: safety, efficacy, and future perspectives
.
Expert Opin Investig Drugs
.
2019 Jan
28
1
73
84
.
43.
Rittchen
S
,
Heinemann
A
.
Therapeutic potential of hematopoietic prostaglandin D2 synthase in allergic inflammation
.
Cells
.
2019 Jun 20
8
6
619
.
44.
Picado
C
,
Fernandez-Morata
JC
,
Juan
M
,
Roca-Ferrer
J
,
Fuentes
M
,
Xaubet
A
.
Cyclooxygenase-2 mRNA is downexpressed in nasal polyps from aspirin-sensitive asthmatics
.
Am J Respir Crit Care Med
.
1999 Jul
160
1
291
6
.
45.
Potaczek
DP
,
Trad
G
,
Sanak
M
,
Garn
H
,
Mastalerz
L
.
Local and systemic production of pro-inflammatory eicosanoids is inversely related to sensitization to aeroallergens in patients with aspirin-exacerbated respiratory disease
.
J Pers Med
.
2022 Mar 11
12
3
447
.
46.
Balzar
S
,
Fajt
ML
,
Comhair
SA
,
Erzurum
SC
,
Bleecker
E
,
Busse
WW
.
Mast cell phenotype, location, and activation in severe asthma. Data from the Severe Asthma Research Program
.
Am J Respir Crit Care Med
.
2011 Feb 1
183
3
299
309
.
47.
Fajt
ML
,
Gelhaus
SL
,
Freeman
B
,
Uvalle
CE
,
Trudeau
JB
,
Holguin
F
.
Prostaglandin D pathway upregulation: relation to asthma severity, control, and TH2 inflammation
.
J Allergy Clin Immunol
.
2013 Jun
131
6
1504
12
.
48.
Wenzel
SE
,
Westcott
JY
,
Larsen
GL
.
Bronchoalveolar lavage fluid mediator levels 5 minutes after allergen challenge in atopic subjects with asthma: relationship to the development of late asthmatic responses
.
J Allergy Clin Immunol
.
1991 Feb
87
2
540
8
.
49.
Okano
M
,
Fujiwara
T
,
Sugata
Y
,
Gotoh
D
,
Masaoka
Y
,
Sogo
M
.
Presence and characterization of prostaglandin D2-related molecules in nasal mucosa of patients with allergic rhinitis
.
Am J Rhinol
.
2006 May–Jun
20
3
342
8
.
50.
Werder
RB
,
Lynch
JP
,
Simpson
JC
,
Zhang
V
,
Hodge
NH
,
Poh
M
.
PGD2/DP2 receptor activation promotes severe viral bronchiolitis by suppressing IFN-lambda production
.
Sci Transl Med
.
2018 May 9
10
440
eaao0052
.
51.
James
KM
,
Gebretsadik
T
,
Escobar
GJ
,
Wu
P
,
Carroll
KN
,
Li
SX
.
Risk of childhood asthma following infant bronchiolitis during the respiratory syncytial virus season
.
J Allergy Clin Immunol
.
2013 Jul
132
1
227
9
.
52.
Santus
P
,
Radovanovic
D
.
Prostaglandin D2 receptor antagonists in early development as potential therapeutic options for asthma
.
Expert Opin Investig Drugs
.
2016 Sep
25
9
1083
92
.
53.
Kupczyk
M
,
Kuna
P
.
Targeting the PGD2/CRTH2/DP1 signaling pathway in asthma and allergic disease: current status and future perspectives
.
Drugs
.
2017 Aug
77
12
1281
94
.
54.
Oguma
T
,
Palmer
LJ
,
Birben
E
,
Sonna
LA
,
Asano
K
,
Lilly
CM
.
Role of prostanoid DP receptor variants in susceptibility to asthma
.
N Engl J Med
.
2004 Oct 21
351
17
1752
63
.
55.
Potaczek
DP
,
Harb
H
,
Michel
S
,
Alhamwe
BA
,
Renz
H
,
Tost
J
.
Epigenetics and allergy: from basic mechanisms to clinical applications
.
Epigenomics
.
2017 Apr
9
4
539
71
.
56.
Potaczek
DP
,
Alashkar Alhamwe
B
,
Miethe
S
,
Garn
H
.
Epigenetic mechanisms in allergy development and prevention
.
Handb Exp Pharmacol
.
2022
;
268
:
331
57
.
57.
Wambre
E
,
DeLong
JH
,
James
EA
,
LaFond
RE
,
Robinson
D
,
Kwok
WW
.
Differentiation stage determines pathologic and protective allergen-specific CD4+ T-cell outcomes during specific immunotherapy
.
J Allergy Clin Immunol
.
2012 Feb
129
2
544
51
, 551.e1-7.
58.
Jaffe
AE
,
Irizarry
RA
.
Accounting for cellular heterogeneity is critical in epigenome-wide association studies
.
Genome Biol
.
2014 Feb 4
15
2
R31
.

Additional information

Edited by: S. Weidinger, Kiel.